Download Urothelial cancers: clinical and imaging evaluation

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Positron emission tomography wikipedia , lookup

Radiosurgery wikipedia , lookup

Neutron capture therapy of cancer wikipedia , lookup

Image-guided radiation therapy wikipedia , lookup

Medical imaging wikipedia , lookup

Transcript
Review Article
Turk J Med Sci
2012; 42 (Sup.2): 1355-1364
© TÜBİTAK
E-mail: [email protected]
doi:10.3906/sag-1111-9
Urothelial cancers: clinical and imaging evaluation
Halil ARSLAN, Fatih Mehmet TEZCAN, Oktay ALĞIN
Abstract: Urothelial cancers are currently diagnosed more commonly parallel to the recent developments in imaging
techniques. Early diagnosis of these tumors, together with better treatment options, decreases the morbidity and the
mortality significantly. This review aims to summarize the imaging findings of urothelial tumors, both at the pretreatment
and posttreatment follow-up stage.
Key words: MRI, urography, hematuria, ureter cancer, urinary tract cancer, computed tomography, diagnosis, staging,
ultrasound, urinary tract imaging, urothelial tumors
Introduction
Urothelial tumors originate from kidney collecting
systems, ureters, the bladder, and the urethra.
Their diagnosis and treatment approaches are quite
different from those of parenchymal tumors of the
kidney (1–3). Radiologists are expected to depict
the presence, location, depth penetration, and local
and distant metastatic status of these lesions, which
usually present with painless hematuria (1–3).
Herein, we aimed to review the radiologic approach,
diagnostic methods, and differential diagnosis of
urothelial tumors.
Urothelial tumors include pelvic, ureteral,
bladder, and urethra tumors, and the majority
of them are bladder tumors (3). They are most
commonly seen in the 6th and 7th decades of life
with a 4:1 male-to-female ratio (1–3). Transitional
cell carcinoma (TCC) constitutes 95% of malignant
urothelial tumors (3–6). More than 90% of all
bladder cancers are TCC, 5%–10% are squamous
cell carcinoma (SCC), and the remaining 2%–3% are
adenocarcinoma (3,6–10). SCC is associated with
chronic inflammation, whereas adenocarcinoma is
associated with persistent urachal remnants (3,11).
Bladder cancers are commonly located in the
trigone and its adjacency (4–10). The second most
common location of TCC is the renal pelvis (1,11).
It has a tendency to be multifocal and of low grade,
similar to in the bladder (1,3). TCC of the urethra
commonly involves the distal portion and tends to be
superficial, but it behaves more aggressively, unlike in
the bladder (4–8). Other rare urinary system tumors
are lipoma, fibroma, leiomyoma, hemangioma,
sarcoma, lymphoma, mesodermal tumors, carcinoid
tumors, and pheochromocytoma, and these should
be considered in differential diagnosis (12).
Etiology
Risk factors for TCC include smoking, analgesics (i.e.
phenacetin), carcinogens (arsenic), schistosomiasis,
familial cancer syndromes, and renal papillary
necrosis (4). Smoking increases the risk 4-fold (13).
Balkan nephropathy, which is seen in Europe, also
increases the risk for TCC (14). TCC tends to be
multifocal and highly recurrent (5,15).
Staging
Pathologic staging
Urothelial tumors are grouped into 2 types, based
on their morphology, as invasive and superficial.
Superficial ones are usually papillary, of low grade,
Received: 13.11.2011 – Accepted: 11.04.2012
Department of Radiology, Atatürk Training and Research Hospital, Bilkent, Ankara – TURKEY
Correspondence: Oktay ALĞIN, Department of Radiology, Atatürk Training and Research Hospital, Bilkent, Ankara – TURKEY
E-mail: [email protected]
1355
Urothelial cancers: clinical and imaging evaluation
and limited to the mucosa lamina propria (3).
They rarely metastasize; however, recurrence after
treatment is common with a good prognosis (5,16).
Invasive ones, however, generally appear smooth
and develop from high-grade in situ lesions; they are
localized to the surface epithelium. They appear as
hyperemic mucosa at cystoscopy (3). Carcinoma in
situ lesions can manifest with tumor cells in urine
cytology and are known as precursors of invasive
cancers (17).
and paraaortic lymph node involvements are also
considered as distant metastases (1).
Renal pelvis tumors are generally malignant; they
involve both kidneys equally and are more commonly
seen in males (8). Well-differentiated tumors are
Grade I and intermediately differentiated ones are
Grade II, whereas poorly differentiated tumors are
Grade III (16).
T2: Muscular layer is invaded by the tumor
(Figure 1b).
Clinical staging
Clinical staging is done according to tumor–node–
metastasis (TNM) classification (3). An analysis
of the depth and spread of the tumor and lymph
node involvement and a metastases workup are
performed (2). Locally, they may involve the bladder
orifice and trigone, ureters, uterus, vagina, urethra,
vagina, prostate, and rectum (3–8,11). Lymphatic
spread occurs through the paravesical, obturator,
external iliac, and paraaortic lymph nodes, whereas
hematogenous spread includes the liver, lungs,
bones, and adrenal glands (2–5,18). Common iliac
Tumor staging (2,3)
Tx: Absence of an evaluable primary tumor.
T0: No tumor.
Tis: Carcinoma in situ.
Ta: Papillary tumor in the epithelium (Figure 1a).
T1: Lamina propria is invaded by the tumor.
T2a: Superficial muscular layer is invaded by the
tumor.
T2b: Deep muscular layer invaded by the tumor.
T3: Perivesical fat planes are invaded.
T3a: Microscopic invasion of the perivesical fat
planes.
T3b: Macroscopic invasion of the perivesical fat
planes.
T4a: Adjacent organs (prostate, rectum, and
vagina) are invaded by the tumor (Figure 2).
T4b: Abdomen–pelvic wall is invaded by the
tumor.
Lymph nodes
Nx: Nodal metastases are unknown.
Figure 1. A patient with papillary bladder cancer (TNM staging: T2b): a) axial T2-weighting MR image demonstrates multiple,
projectile, papillary masses in the bladder, and b) axial diffusion-weighted image also localizes these masses with a
hyperintense signal pattern.
1356
H. ARSLAN, F. M. TEZCAN, O. ALĞIN
Figure 2. Axial CT images of a bladder cancer patient (T4
disease with seminal vesicle invasion) shows multiple
focal wall thickening and a right posterolaterally
localized mass (arrows) extruding from the wall
into the lumen with contrast material enhancement
(arrows) (upper image). The perivesical fat planes
and perirectal space are both invaded by the lesion
(arrows) (lower image).
Figure 3. Axial CT images of a bladder cancer patient show
a right bladder wall mass (star) (upper image) with
enhancing perivesical and perirectal lymph nodes
(arrows), which correspond to N2 disease (lower
image).
Imaging techniques for diagnosis
and magnetic resonance imaging (MRI). The role
of the direct urinary system radiograph is limited
to depicting the presence of calcification and gas
in the collecting system (2). IVP is less sensitive in
the detection of smaller lesions (10). US is usually
used as a first-line method and is sensitive in the
detection of bladder tumors. US can be performed
by transabdominal, transrectal, or transurethral
approaches. CT is the most commonly used method
for evaluation of the primary tumor, lymph nodes,
and metastases (1). MRI is more sensitive than CT
due to its superior soft tissue contrast resolution
and it is useful in the investigation of tumoral depth
penetration, distant metastases, bony involvement,
and late fibrosis versus recurrence distinction (19–
22).
Conventional cystoscopy is the gold standard of
diagnostic methods (1–3). Imaging methods for
diagnosis include intravenous pyelography (IVP),
ultrasound (US), computed tomography (CT),
IVP is the traditional initial imaging method in the
imaging workup of hematuria (2). At the initial step
of IVP, a direct urinary system radiograph should be
N1: Single pelvic lymph node less than 2 cm in
diameter (Figure 3a).
N2: Pelvic lymph node metastases with a diameter
of 2 cm to 5 cm (Figure 3b).
N3: Pelvic lymph node metastases greater than 5
cm in diameter.
Metastases
MX: Distant metastases are unknown.
M0: Absence of distant metastases.
M1: Presence of distant metastases.
Intravenous pyelography
1357
Urothelial cancers: clinical and imaging evaluation
obtained prior to the contrast injection in order to
visualize any opacity (10). IVP has nephrography,
pyelography, and cystography phases (2). Large
tumors appear as filling defects at the pyelography
and cystography phases (23), whereas small tumors
and intradiverticular lesions may not be visualized
via this imaging method (10). The main limitation of
this method is the radiation exposure. Additionally,
as it includes an intravenous iodine-based contrast
agent, it is contraindicated in patients with kidney
failure and contrast allergy history (2,21,23). IVP has
an accuracy range of 26% to 87% in the detection of
bladder cancers with a smallest detected lesion size
cutoff of 1.5 cm (2,9,10,23).
Ultrasound
US is a relatively cheaper and more accessible imaging
method and is used as a first-line technique for the
imaging workup of hematuria. US can precisely depict
bladder and pelvis tumors in experienced hands but
is limited in ureteral lesions due to gas superposition
(2). Hydronephrosis of the kidneys, stones, cysts, and
other filling defects within kidney collecting systems
can be easily demonstrated at US, whereas bladder
lesions appear as a projectile mass or wall thickening
(2) (Figure 4). The disruption of the echogenic line
around the bladder wall at US represents a possible
invasion in bladder cancer patients (23).
A relatively less invasive nature, lack of any
requirement for an intravenous contrast agent, and
repeatability are among the main advantages of US;
however, operator dependence, difficulty in cases
of patient obesity, and abdominal gas superposition
are among the main disadvantages of this technique.
Bladder lesions that are smaller than 5 mm and
localized to the bladder dome or neck can be missed
via US (2,23). The overall accuracy of US for bladder
cancer detection is around 82%–95% regardless of the
lesion size (24,25). US can be limited in visualization
of the distal renal pelvis, where IVP and CT have
more success, but hydronephrosis secondary to
tumors can be detected and graded by US (9,10).
Additionally, US can be helpful in the evaluation of
patients with sepsis, when IVP is contraindicated.
Finally, Kocakoc et al. recently reported that virtual
(3-dimensional) ultrasonographic cystoscopy can
be a useful alternative for the evaluation of bladder
cancers (26).
1358
Figure 4. Axial ultrasound image of a patient with papillary
carcinoma shows a projectile mass in the left bladder
wall (arrow).
Computed tomography
CT is the most commonly used imaging method
for urinary system tumors, especially in staging
(9,10). The advent of multidetector technology has
increased its use and utility in the evaluation of
urinary system tumors. A standard abdomen CT
scan includes an intravenous injection of 100–120
mL of nonionic contrast material, at an injection rate
of 2.5–3 mL/s and a subsequent image acquisition
at the parenchymal phase, with an approximate
delay of 60–100 s following the injection (2,27).
Kidney parenchyma and tumor enhancement can be
evaluated at the nephrographic phase (9,10) (Figure
5). Epithelial tumors can be assessed more accurately
at the excretory or pyelographic phase and they
appear as filling defects (1,2) (Figure 6). The bladder
should be sufficiently full and distended for proper
lesion evaluation (Figure 7). Reconstructed images
in various planes enable better evaluation of lesions
localized to the dome and base of the bladder (27)
(Figure 7). In some patients, ureteral lesions can be
barely visualized, especially if there is incomplete
opacification (9,10). In addition to routine CT scans,
several special methods can also be used. These are
summarized below.
CT urography
CT urography is the cross-sectional analog of IVP.
In this technique, image acquisition is performed at
7–15 min after the intravenous contrast injection and
H. ARSLAN, F. M. TEZCAN, O. ALĞIN
Figure 5. Axial CT image of a patient with ureteral carcinoma
shows a nodular wall thickening of the distal right
ureter with heterogeneous contrast enhancement and
luminal narrowing (arrow).
the images are usually evaluated after a maximum
intensity projection (MIP) reconstruction algorithm
application (9,10). Tumors appear as focal wall
thickening or as masses projecting into the lumen
(9,10). The usage of multidetector scanners increases
both the sensitivity of this technique as well as
enables a more accurate depiction of lesions smaller
than 4 mm (28).
Antegrade CT pyelography
In this technique, the renal pelvis is imaged following
catheterization after a subsequent contrast injection
through the catheter. In patients with kidney failure,
this technique allows for the determination of the
obstruction site without an intravenous contrast
injection (2). Moreover, this technique can be
considered as an alternative in patients with contrast
allergy (Figure 8) (29). However, the invasiveness of
this technique is a major drawback.
CT cystoscopy and virtual cystoscopy
This technique enables images to be obtained that
are similar to those of conventional cystoscopy.
However, its major limitations are the lack of realtime tissue sampling (biopsy), the inability to evaluate
mucosa and carcinoma in situ, and the less accurate
demonstration of very small lesions (23,30,31).
On the other hand, it has some advantages over
conventional cystoscopy, such as the fact that it can
be easily performed as a noninvasive alternative in
patients with benign prostate hyperplasia, bladder
infections, urethral strictures, and prostatitis, when
the conventional technique is contraindicated (32).
Figure 6. Axial CT image at the pyelography phase shows a
hypodense filling defect in the right renal pelvis,
which is consistent with a TCC (star).
Additionally, virtual cystoscopy is more successful in
the evaluation of the bladder neck and in diverticula
with a narrow neck, where the conventional
technique is limited (32,33). The virtual cystoscopy
method allows for a 360° visualization of the bladder
on all planes, similar to or even better than the
conventional method (32). Through this technique,
the location and texture of the lesion can be assessed
prior to biopsy or surgery, and this can aid in surgical
planning and reduce the procedure time. Moreover, it
can be used at the follow-up, as well (30–33) (Figure
9).
In this technique, the bladder is filled directly with
air (minimally invasive, through a Foley catheter)
or indirectly with intravenous contrast material
(noninvasively). The air-filled approach requires
2 CT data sets (prone and supine) and therefore
has a higher radiation dose exposure. The indirect
method is performed at a supine position with a
lower radiation dose exposure (34). However, in the
indirect approach, virtual images can be obscured
by artifacts secondary to the improper mixture of
the contrast material and urine, especially in the
increased trabeculation areas of the bladder wall (35).
Magnetic resonance imaging
MR urography
MR urography (MRU) can be used for patients
with an iodine-based contrast material allergy or
minimal renal insufficiency instead of CT urography
1359
Urothelial cancers: clinical and imaging evaluation
Figure 7. Sagittal (left) and coronal (right) reconstructed CT
images show multiple, projectile enhancing tumor
lesions in the bladder (arrows).
(20,21,36). Under such circumstances, it is safer to
use macrocyclic MRI contrast agents (21). MRU has
both static and functional components. In the static
part (also called a noncontrast-enhanced or static
MRU), heavily T2-weighted sequences are obtained
without a contrast material injection, whereas in
the functional part (also called a contrast materialenhanced or excretory MRU), the contrast-enhanced
3D gradient echo (GRE) T1-weighted sequences are
acquired (20,37–39). Noncontrast-enhanced MRU
(NCE-MRU) has less sensitivity for the detection of
smaller lesions (37).
In contrast-enhanced MRU (CE-MRU),
intravenous furosemide (10–20 mg) can be used to
avoid the T2* effect of the contrast dilution and to
distend the urinary system sufficiently (40). The most
important postcontrast T1-weighted images are those
obtained at the nephrographic and pyelographic
phases. Smaller lesions appear as enhancing foci at
the nephrographic phase, whereas they appear as
a filling defect at the delayed excretory phase (36).
Ureteral lesions can be visualized in either of these
sequences (10).
Papillary tumors are seen as filling defects at
MRU (36). Postcontrast T1-weighted images at the
nephrographic phase are helpful for diagnosis and
such lesions can be differentiated from others by
their uniform homogeneous contrast enhancement
1360
Figure 8. MIP image of an antegrade CT pyelogram of S/P
surgery bladder cancer patient obtained following
contrast injection through bilateral nephrostomy
catheters.
(41). Flat lesions are challenging to depict due to
hyperintense urine on T2-weighted sequences or
at NCE-MRU and can be easily missed. CE-MRU
is more useful in the diagnosis of such lesions
(10,40,41).
MR cystography and virtual MR cystoscopy
About 80% of bladder cancers are polypoid, and
MR cystography and virtual MR cystoscopy are
important in localizing such lesions (41). On T2weighted images, since urine appears hyperintense,
it is easier to visualize pathologies with different
signal characteristics. Virtual cystoscopy is obtained
through the postprocessing of the T2-weighted data
with multiplanar reconstruction and MIP algorithms.
MR cystoscopy has higher sensitivity and specificity
for lesions greater than 10 mm, whereas these values
diminish for lesions smaller than 10 mm. Virtual
cystoscopy performed with multidetector CT has
a better spatial resolution than that of MRI, and
therefore it is more sensitive to smaller lesions. Mural
H. ARSLAN, F. M. TEZCAN, O. ALĞIN
Figure 9. CT cystography (left) and virtual cystoscopy (right) images demonstrate a polypoid projectile lesion in the left lateral bladder
wall (arrows). These figures reprinted permission from reference 31.
lesions and perivesical infiltration cannot be precisely
detected at MR cystography. Moreover, in situ lesions
cannot be identified, as there is no information about
mucosal color changes (42,43).
The main advantages in using this technique as
an alternative in case of a contraindication for the
conventional cystoscopy are better localization of the
bladder neck and intradiverticular lesions and the
easiness of the follow-up of patients with local excision
(44). The main disadvantages are operator dependency,
inability to obtain real-time biopsy, higher cost, and
the requirement of higher-strength magnet systems
(42). The common indications for MR cystoscopy can
be summarized as the need for bladder evaluation in
case of urethral strictures, the investigation of bladder
diverticula, and conditions in which conventional
cystoscopy is contraindicated (42).
Diffusion-weighted MRI
Diffusion refers to the motion of the water molecules
in biologic tissues following excitation via heat, which
is also known as Brownian motion (45–47). Cancers
appear as hyperintense foci on diffusion-weighted
MRI (DW MRI) whereas they are hypointense on
apparent diffusion coefficient (ADC) maps derived
from DW MRI, and this may help to distinguish
low-grade lesions from high-grade ones (48). ADC
is a reliable quantitative measure and ADC values
can help to differentiate cancers from cystitis or wall
hypertrophy secondary to outlet obstruction (45–47).
Differential diagnosis
Filling defects, which are among the most common
imaging features of urothelial tumors, can also be
seen in other pathologies that also result in diffuse
enhancement, wall thickening, and focal infiltration
Figure 10. CE-MRU and multidetector CT images of 3 different patients. Coronal postcontrast 3D T1-weighting gradient-echo MR
image (CE-MRU) in a patient with ileal conduit shows a patent passage (arrows in left image). Coronal CE-MRU image of a
different patient shows a leak from the anastomoses (arrow in middle image). Right image shows bladder perforation at the
postoperative stage.
1361
Urothelial cancers: clinical and imaging evaluation
(10). Stone, clot, mucous, debris, fungus ball,
concentrated gadolinium, flow-related artifacts,
vascular partial volume, inflammation, reflux,
peristaltic motion, and jet flow should be considered
in the differential diagnosis of urothelial carcinoma
(35–37,49–51). Additionally, nephrostomy tubes,
instrumentations, urinary stasis, intravesical
chemotherapy agents, neobladder and urinary
diversions, and urinary stents can mimic diffuse
contrast enhancement, as well (1,2).
Lymph node metastasis
Spherical lymph nodes greater than 8 mm, oval
or ellipsoid nodes greater than 10 mm with rapid
enhancement at dynamic scans, and clustered
lymph nodes are important findings for metastatic
involvement (1–4,52). A few publications have
reported that ADC values can be helpful in
differentiating benign nodes from malignant ones;
however, further research is required to explore this
topic (53).
Surgical treatment of bladder tumors
Treatment approach varies with patient age and
comorbidities. The aim of the treatment in superficial
(stage T1) lesions is to prevent recurrence or
progression. A bladder-sparing treatment is applied
in early-stage tumors, and partial cystectomy or
transurethral tumor resections are among such
techniques (54). In situ lesions tend to be high
grade and they may require cystectomy if there
is no response to 2 episodes of bacillus CalmetteGuérin treatment for 6 weeks. T2 and T3 lesions
usually require cystectomy as they have infiltrative
characteristics in the presence of nodal infiltration,
and chemotherapy and radiotherapy should be
added to the treatment regimen. Cystectomy is a
well-known standard treatment for patients with
invasive tumor (54,55). After pelvic surgery, bleeding
is a common and significant risk. Techniques of
interventional radiology are noninvasive and safe
methods for demonstration of the bleeding area and
effective treatment (56).
An artificial bladder is reconstructed in cystectomy
and this is known as a diversion procedure. Several
diversion techniques exist:
Urinary diversions
They are mainly grouped into 2 types.
1362
Noncontinent diversions (ureterostomy and
conduits): In ureterostomy, ureters are directly
attached to the skin without an intestinal loop. This
includes 2 stomas and a pouch. This technique is
rarely used. In case of an ileal conduit, a reservoir
is created from a short segment of the small bowel
and the ureters are attached to one side of this loop,
whereas the other side is attached to the skin to make
up the stoma (Figure 10). An additional pouch is also
created in the opening of the stoma. This technique
is well established with long-term complication and
collecting system dilatation rates of 30% and 20%,
respectively (57).
Continent diversions: These are grouped into
2 types: heterotopic and orthotopic. Heterotopic
diversion operations are not performed commonly
nowadays. The most common of them includes
stomatization of the ureters to the sigmoid colon,
but this increases the infection rate. Orthotopic
diversions are more commonly preferred recently
and a new bladder is formed using a small bowel
loop, which replaces the excised bladder. Ureters
are connected to this newly formed bladder and
the patient can perform urinary excretion properly
(58) (Figure 10). Complications such as perforation,
fistula formation, bleeding, and urinoma can be seen
at the postoperative phase (54–58) (Figure 10).
Conclusion
Urothelial cancers usually present with painless
hematuria and are currently more commonly
diagnosed secondary to recent developments in the
imaging technology. The application of an accurate
imaging algorithm to further investigate painless
hematuria will enable prompt and correct diagnosis
of urothelial cancers, which will increase the yield
of treatment and significantly decrease cancer and
treatment-related morbidity and mortality. Moreover,
the angiographic techniques can be useful in the preand postoperative period management of patients
with urothelial cancer.
Acknowledgments
The authors thank Dr Barış Türkbey, Dr Gökçe
Akgündüz, and Dr E. Bengi Türkbey for their
contributions.
H. ARSLAN, F. M. TEZCAN, O. ALĞIN
References
1.
Lee EK, Dickstein RJ, Kamta AM. Imaging of urothelial cancers:
what the urologist needs to know. AJR Am J Roentgenol 2011;
196: 1249–54.
18. Greene FL, Compton CC, Fritz AG, Shah JP, Winchester DP.
AJCC cancer staging atlas. Berlin, Germany: Springer-Verlag;
2006.
2.
Gödekmerdan A, Yahşi S, Semerciöz A, İlhan F, Akpolat N,
Yekeler H. Determination of nuclear matrix protein 22 levels
in cystitis, urothelial dysplasia and urothelial carcinoma. Turk
J Med Sci 2006; 36: 93–6.
19. Cowan NC, Crew JP. Imaging bladder cancer. Curr Opin Urol
2010; 20: 409–13.
20. Algin O. Functional evaluation of kidneys: which technique is
more useful? AJR Am J Roentgenol 2010; 194: W241.
3.
Wong-You-Cheong JJ, Woodward PJ, Manning MA, Sesterhenn
IA. From the archives of the AFIP: neoplasms of the urinary
bladder: radiologic-pathologic correlation. Radiographics
2006; 26: 553–80.
4.
Ng CS. Radiologic diagnosis and staging of renal and bladder
cancer. Semin Roentgenol 2006; 41: 121–38.
22. Türkbey B, Bernardo M, Merino MJ, Wood BJ, Pinto PA,
Choyke PL. MRI of localized prostate cancer: coming of age in
the PSA era. Diagn Interv Radiol 2012; 18: 34–45.
5.
Vikram R, Sandler CM, Ng CS. Imaging and staging of
transitional cell carcinoma. Part 1. Lower urinary tract. AJR
Am J Roentgenol 2009; 192: 1481–7.
23. Rafique M, Javed AA. Role of intravenous urography and
transabdominal ultrasonography in the diagnosis of bladder
carcinoma. Int Braz J Urol 2004; 30: 185–91.
6.
Vikram R, Sandler CM, Ng CS. Imaging and staging of
transitional cell carcinoma. Part 2. Upper urinary tract. AJR
Am J Roentgenol 2009; 192: 1488–93.
7.
Browne RF, Meehan CP, Colville J, Power R, Torreggiani
WC. Transitional cell carcinoma of the upper urinary tract:
spectrum of imaging findings. Radiographics 2005; 25: 1609–
27.
24. Malone PR, Weston-Underwood J, Aron PM, Wilkinson KW,
Joseph AE, Riddle PR. The use of transabdominal ultrasound
in the detection of early bladder tumours. Br J Urol 1986; 58:
520–2.
8.
Lakadamyalı H, Akata D, Akdoğan B, Akhan O. Multicentric
uroepithelial tumor: CT and MRI findings. Diagn Intervent
Radiol 2002; 8: 85–9.
9.
Caoili EM, Cohan RH, Inampudi P, Ellis JH, Shah RB, Faerber
GJ et al. MDCT urography of upper tract urothelial neoplasms.
AJR Am J Roentgenol 2005; 184: 1873–81.
10. Akbulut Z, Tuzlalı M, Canda AE, Ercan K, Kandemir O, Balbay
MD. Factors affecting adrenal gland involvement in patients
who underwent radical nephrectomy for renal cell carcinoma.
Turk J Med Sci 2009; 39: 215–22.
11. Rafla S. Tumors of the upper urothelium. Am J Roentgenol
Radium Ther Nucl Med 1975; 123: 540–51.
12. Mostofi FK: Histological typing of urinary bladder tumours.
Berlin: Springer-Verlag; 1999. p.3–27.
13. Bostwick DG, Ramnani D, Cheng L. Diagnosis and grading
of bladder cancer and associated lesions. Urol Clin North Am
1999; 26: 493–507.
14. Stefanovic V, Radovanovic Z. Balkan endemic nephropathy
and associated urothelial cancer. Nat Clin Pract Urol 2008; 5:
105–12.
15. Elmajian DA. Transitional cell carcinoma of the ureter and
renal pelvis. In: Nachtsheim D, editor. Urological oncology.
Austin (TX): Landes Bioscience; 2005. p.43–52.
16. Fleshner N, Kondylis F. Demographics and epidemiology of
urothelial cancer of urinary bladder. In: Droller MJ, editor.
Urothelial tumors. Hamilton (ON): Decker; 2004. p.1–16.
17. Pashos CL, Botteman MF, Laskin BL, Redaelli A. Bladder
cancer: epidemiology, diagnosis, and management. Cancer
Pract 2002; 10: 311–22.
21. Algin O. A new contrast media for functional MR urography:
Gd-MAG3. Med Hypotheses 2011; 77: 74–6.
25. Itzchak Y, Singer D, Fischelovitch Y. Ultrasonographic
assessment of bladder tumors. I. Tumor detection. J Urol 1981;
126: 31–3.
26. Kocakoc E, Kiris A, Orhan I, Poyraz AK, Artas H, Firdolas F.
Detection of bladder tumors with 3-dimensional sonography
and virtual sonographic cystoscopy. J Ultrasound Med 2008;
27: 45–53.
27. Ozbülbül NI, Dağli M, Akdoğan G, Olçer T. CT urography of
a vesicourachal diverticulum containing calculi. Diagn Interv
Radiol 2010; 16: 56–8.
28. Wang LJ, Wong YC, Ng KF, Chuang CK, Lee SY, Wan YL.
Tumor characteristics of urothelial carcinoma on multidetector
computerized tomography urography. J Urol 2010; 183: 2154–
60.
29. Ghersin E, Brook OR, Meretik S, Kaftori JK, Ofer A, Amendola
MA et al. Antegrade MDCT pyelography for the evaluation of
patients with obstructed urinary tract. AJR Am J Roentgenol
2004; 183: 1691–6.
30. Song JH, Francis IR, Platt JF, Cohan RH, Mohsin J, Kielb SJ
et al. Bladder tumor detection at virtual cystoscopy. Radiology
2001; 218: 95–100.
31. Arslan H, Ceylan K, Harman M, Yilmaz Y, Temizoz O, Can
S. Virtual computed tomography cystoscopy in bladder
pathologies. Int Braz J Urol 2006; 32: 147–54.
32. Tsampoulas C, Tsili AC, Giannakis D, Alamanos Y, Sofikitis
N, Efremidis SC. 16-MDCT cystoscopy in the evaluation of
neoplasms of the urinary bladder. AJR Am J Roentgenol 2008;
190: 729–35.
33. Frank R, Stenzl A, Frede T, Eder R, Recheis W, Knapp R et al.
Three-dimensional computed tomography of the reconstructed
lower urinary tract: technique and findings. Eur Radiol 1998; 8:
657–63.
1363
Urothelial cancers: clinical and imaging evaluation
34. Kim JK, Park SY, Kim HS, Kim SH, Cho KS. Comparison of
virtual cystoscopy, multiplanar reformation, and source CT
images with contrast material-filled bladder for detecting
lesions. AJR Am J Roentgenol 2005; 185: 689–96.
47. Shimofusa R, Fujimoto H, Akamata H, Motoori K, Yamamoto
S, Ueda T et al. Diffusion-weighted imaging of prostate cancer.
J Comput Assist Tomogr 2004; 29: 149–53.
35. Nambirajan T, Sohaib SA, Muller-Pollard C, Reznek R,
Chinegwundoh FI. Virtual cystoscopy from computed
tomography: a pilot study. BJU Int 2004; 94: 828–31.
48. Türkbey B, Aras Ö, Karabulut N, Turgut AT, Akpınar E, Alibek
S et al. Diffusion-weighted MRI for detecting and monitoring
cancer: a review of current applications in body imaging.
Diagn Interv Radiol 2012; 18: 46–59.
36. Takahashi N, Kawashima A, Glockner JF, Hartman RP, Kim B,
King BF. MR urography for suspected upper tract urothelial
carcinoma. Eur Radiol 2009; 19: 912–23.
49. Blandino A, Gaeta M, Minutoli F, Salamone I, Magno C,
Scribano E et al. MR urography of the ureter. AJR Am J
Roentgenol 2002; 179: 1307–14.
37. Takahashi N, Kawashima A, Glockner JF, Hartman RP,
Leibovich BC, Brau AC et al. Small (<2-cm) upper-tract
urothelial carcinoma: evaluation with gadolinium-enhanced
three-dimensional spoiled gradient-recalled echo MR
urography. Radiology 2008; 247: 451–7.
50. Heneghan JP, Dalrymple NC, Verga M, Rosenfield AT, Smith
RC. Soft-tissue “rim” sign in the diagnosis of ureteral calculi
with use of unenhanced helical CT. Radiology 1997; 202: 709–
11.
38. Broome DR, Girguis MS, Baron PW, Cottrell AC, Kjellin I, Kirk
GA. Gadodiamide-associated nephrogenic systemic fibrosis:
why radiologists should be concerned. AJR Am J Roentgenol
2007; 188: 586–92.
39. Morcos SK. Extracellular gadolinium contrast agents:
differences instability. Eur J Radiol 2008; 66: 175–9.
40. Nolte-Ernsting CC, Bücker A, Adam GB, Neuerburg JM, Jung
P, Hunter DW et al. Gadolinium-enhanced excretory MR
urography after low-dose diuretic injection: comparison with
conventional excretory urography. Radiology 1998; 209: 147–
57.
41. Merkle EM, Wunderlich A, Aschoff AJ, Rilinger N, Görich J,
Bachor R et al. Virtual cystoscopy based on helical CT scan
datasets: perspectives and limitations. Br J Radiol 1998; 71:
262–7.
51. Chahal R, Taylor K, Eardley I, Lloyd SN, Spencer JA. Patients
at high risk for upper tract urothelial cancer: evaluation of
hydronephrosis using high resolution magnetic resonance
urography. J Urol 2005; 174: 478–82.
52. Jager GJ, Barentsz JO, Oosterhof GO, Witjes JA, Ruijs SJ. Pelvic
adenopathy in prostatic and urinary bladder carcinoma: MR
imaging with a three-dimensional TI-weighted magnetizationprepared-rapid gradient-echo sequence. AJR Am J Roentgenol
1996; 167: 1503–7.
53. Chen YB, Liao J, Xie R, Chen GL, Chen G. Discrimination of
metastatic from hyperplastic pelvic lymph nodes in patients
with cervical cancer by diffusion-weighted magnetic resonance
imaging. Abdom Imaging 2011; 36: 102–9.
54. Oosterlinck W, Lobel B, Jakse G, Malmström PU, Stöckle M,
Sternberg C et al. Guidelines on bladder cancer. Eur Urol 2002;
41: 105–12.
42. Beer A, Saar B, Zantl N, Link TM, Roggel R, Hwang SL et al.
MR cystography for bladder tumor detection. Eur Radiol 2004;
14: 2311–9.
55. Cookson MS, Herr HW, Zhang ZF, Soloway S, Sogani PC, Fair
WR. The treated natural history of high risk superficial bladder
cancer: 15-year outcome. J Urol 1997; 158: 62–7.
43. Fenlon HM, Bell TV, Ahari HK, Hussain S. Virtual cystoscopy:
early clinical experience. Radiology 1997; 205: 272–5.
56. Karaman B, Oren NC, Andic C, Ustunsoz B. Transcatheter
embolization for the treatment of both vaginal and lower
intestinal bleeding due to advanced pelvic malignancy.
Eurasian J Med 2010; 42: 153–6.
44. Läemmle M, Beer A, Settles M, Hannig C, Schwaibold H,
Drews C. Reliability of MR imaging based virtual cystoscopy
in the diagnosis of cancer of the urinary bladder. AJR Am J
Roentgenol 2002; 178: 1483–8.
45. Matsuki M, Inada Y, Tatsugami F, Tanikake M, Narabayashi
I, Katsuoka Y. Diffusion-weighted MR imaging for urinary
bladder carcinoma: initial results. Eur Radiol 2007; 17: 201–4.
46. Takahara T, Imai Y, Yamashita T, Yasuda S, Nasu S, Van
Cauteren M. Diffusion weighted whole body imaging with
background body signal suppression (DWIBS): technical
improvement using free breathing, STIR and high resolution
3D display. Radiat Med 2004; 22: 275–82.
1364
57. Neal DE. Complications of ileal conduit diversion in adults
with cancer followed up for at least five years. Br Med J (Clin
Res Ed) 1985; 290: 1695–7.
58. Hautmann RE, de Petriconi R, Gottfried HW, Kleinschmidt
K, Mattes R, Paiss T. The ileal neobladder: complications and
functional results in 363 patients after 11 years of follow up. J
Urol 1999; 161: 422–7.